Download as pdf or txt
Download as pdf or txt
You are on page 1of 5

Colloquium

⌬FosB: A sustained molecular switch for addiction


Eric J. Nestler*, Michel Barrot, and David W. Self
Department of Psychiatry and Center for Basic Neuroscience, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard,
Dallas, TX 75390-9070

The longevity of some of the behavioral abnormalities that char- and Fra2 (7). These Fos family proteins heterodimerize with Jun
acterize drug addiction has suggested that regulation of neural family proteins (c-Jun, JunB, or JunD) to form active AP-1
gene expression may be involved in the process by which drugs of (activator protein-1) transcription factors that bind to AP-1 sites
abuse cause a state of addiction. Increasing evidence suggests that (consensus sequence: TGAC兾GTCA) present in the promoters
the transcription factor ⌬FosB represents one mechanism by which of certain genes to regulate their transcription.
drugs of abuse produce relatively stable changes in the brain that These Fos family proteins are induced rapidly and transiently
contribute to the addiction phenotype. ⌬FosB, a member of the Fos in specific brain regions after acute administration of many drugs
family of transcription factors, accumulates within a subset of of abuse (Fig. 1) (8–11). Prominent regions are the nucleus
neurons of the nucleus accumbens and dorsal striatum (brain accumbens and dorsal striatum, which are important mediators
regions important for addiction) after repeated administration of of behavioral responses to the drugs, in particular, their reward-
many kinds of drugs of abuse. Similar accumulation of ⌬FosB
ing and locomotor-activating effects (12, 13). These proteins
occurs after compulsive running, which suggests that ⌬FosB may
return to basal levels within hours of drug administration.
accumulate in response to many types of compulsive behaviors.
Very different responses are seen after chronic administration
Importantly, ⌬FosB persists in neurons for relatively long periods
of time because of its extraordinary stability. Therefore, ⌬FosB
of drugs of abuse (Fig. 1). Biochemically modified isoforms of
represents a molecular mechanism that could initiate and then ⌬FosB (molecular mass 35–37 kDa) accumulate within the same
sustain changes in gene expression that persist long after drug brain regions after repeated drug exposure, whereas all other
exposure ceases. Studies in inducible transgenic mice that overex- Fos family members show tolerance (that is, reduced induction
press either ⌬FosB or a dominant negative inhibitor of the protein compared with initial drug exposures). Such accumulation of
provide direct evidence that ⌬FosB causes increased sensitivity to ⌬FosB has been observed for cocaine, morphine, amphetamine,
the behavioral effects of drugs of abuse and, possibly, increased alcohol, nicotine, and phencyclidine (11, 14–18). There is some
drug seeking behavior. This work supports the view that ⌬FosB evidence that this induction is selective for the dynorphin兾
functions as a type of sustained ‘‘molecular switch’’ that gradually substance P-containing subset of medium spiny neurons located
converts acute drug responses into relatively stable adaptations in these brain regions (15, 17), although more work is needed to
that contribute to the long-term neural and behavioral plasticity establish this with certainty. The 35- to 37-kDa isoforms of
that underlies addiction. ⌬FosB dimerize predominantly with JunD to form an active and
long-lasting AP-1 complex within these brain regions (19, 20).
These ⌬FosB isoforms accumulate with chronic drug exposure
A ddiction research is focused on understanding the complex
ways in which drugs of abuse change the brain to cause
behavioral abnormalities that characterize addiction. One of the
because of their extraordinarily long half-lives (21), and there-
fore persist in the neurons for at least several weeks after
critical challenges in the field is to identify relatively stable cessation of drug administration. It is interesting to note that
drug-induced changes in the brain to account for those behav- these ⌬FosB isoforms are highly stable products of an immediate
ioral abnormalities that are particularly long-lived. For example, early gene ( fosB). The stability of the ⌬FosB isoforms provides
a human addict may be at increased risk for relapse even after a novel molecular mechanism by which drug-induced changes in
years of abstinence. gene expression can persist despite relatively long periods of
The stability of these behavioral abnormalities has led to the drug withdrawal.
suggestion that they may be mediated, at least in part, through Although the nucleus accumbens plays a critical role in the
changes in gene expression (1–3). According to this view, rewarding effects of drugs of abuse, it is believed to function
repeated exposure to a drug of abuse repeatedly perturbs normally by regulating responses to natural reinforcers, such as
transmission at particular synapses in the brain that are sensitive food, drink, sex, and social interactions (12, 13). As a result, there
to the drug. Such perturbations eventually signal via intracellular is considerable interest in a possible role of this brain region in
messenger cascades to the nucleus, where they first initiate and other compulsive behaviors (e.g., pathological overeating, gam-
then maintain changes in the expression of specific genes. A bling, exercise, etc.). For this reason, we examined whether
primary mechanism through which signal transduction pathways ⌬FosB is regulated in an animal model of compulsive running.
influence gene expression is the regulation of transcription Indeed, the stable 35- to 37-kDa isoforms of ⌬FosB are induced
factors, proteins that bind to regulatory regions of genes and
selectively within the nucleus accumbens in rats that show
modify their transcription.
compulsive running behavior.†
One goal of addiction research, therefore, has been to identify
transcription factors that are altered in brain regions implicated
in addiction after chronic administration of drugs of abuse. This paper was presented at the Inaugural Arthur M. Sackler Colloquium of the National
Several such transcription factors have been identified over the Academy of Sciences, ‘‘Neural Signaling,’’ held February 15–17, 2001, at the National
past decade (1–6). The focus of this review is on one particular Academy of Sciences in Washington, DC.
transcription factor called ⌬FosB. Abbreviations: AP-1, activator protein-1; AMPA, ␣-amino-3-hydroxy-5-methyl-4-isox-
azolepropionic acid; CREB, cAMP response element binding protein; Cdk5, cyclin-depen-
Induction of ⌬FosB by Drugs of Abuse dent kinase-5.

⌬FosB, encoded by the fosB gene, is a member of the Fos family *To whom reprint requests should be addressed. E-mail: eric.nestler@utsouthwestern.edu.
of transcription factors, which also include c-Fos, FosB, Fra1, †Werme, M., Nestler, E. J. & Brene, S. (2001) Soc. Neurosci. Abstr., in press.

11042–11046 兩 PNAS 兩 September 25, 2001 兩 vol. 98 兩 no. 20 www.pnas.org兾cgi兾doi兾10.1073兾pnas.191352698


Table 1. Behavioral plasticity mediated by ⌬FosB in nucleus
accumbens-dorsal striatum
Increased locomotor activation in response to acute and repeated
cocaine administration.
Increased rewarding responses to cocaine and morphine in
place-conditioning assays.
Increased self-administration of low doses of cocaine.
Increased motivation for cocaine in progressive ratio assays.
Increased anxiolytic responses to alcohol.
Increased compulsive running behavior.

Based on data in refs. 28 and 29.†‡§¶

from the 33-kDa isoform of ⌬FosB that is induced rapidly but


transiently after a single drug exposure (Fig. 1) (14, 19, 22).
Current evidence suggests that the 33-kDa isoform is the native
form of the protein, which is altered to form the more stable 35-
to 37-kDa products (19, 21). However, the nature of the bio-
chemical modification that converts the unstable 33-kDa isoform
into the stable 35- to 37-kDa isoforms has remained obscure. It
has been speculated that phosphorylation may be responsible
(11). For example, induction of ⌬FosB is attenuated in mice
lacking DARPP-32, a striatal-enriched protein (23, 24). Because
DARPP-32 regulates the catalytic activity of protein phospha-
tase-1 and protein kinase A (25, 26), the requirement for this
protein for the normal accumulation of the stable ⌬FosB
isoforms suggests a possible role for phosphorylation in gener-
ation of these stable products.

Role of ⌬FosB in Behavioral Plasticity to Drugs of Abuse


Insight into the role of ⌬FosB in drug addiction has come largely
from the study of transgenic mice in which ⌬FosB can be induced
selectively within the nucleus accumbens and other striatal
regions of adult animals (27, 28). Importantly, these mice
overexpress ⌬FosB selectively in the dynorphin兾substance P-
containing medium spiny neurons, where the drugs are believed
to induce the protein. The behavioral phenotype of the ⌬FosB-
overexpressing mice, which in many ways resembles animals
after chronic drug exposure, is summarized in Table 1. The mice

COLLOQUIUM
show augmented locomotor responses to cocaine after acute and
chronic administration (28). They also show enhanced sensitivity
to the rewarding effects of cocaine and morphine in place-
conditioning assays (11, 28) and will self-administer lower doses
of cocaine than littermates that do not overexpress ⌬FosB.‡ In
contrast, these animals show normal conditioned locomotor
sensitization to cocaine and normal spatial learning in the Morris
water maze (28). These data indicate that ⌬FosB increases an
animal’s sensitivity to cocaine and perhaps other drugs of abuse
Fig. 1. Scheme showing the gradual accumulation of ⌬FosB versus the rapid
and may represent a mechanism for relatively prolonged sensi-
and transient induction of other Fos family proteins in response to drugs of abuse.
(A) The autoradiogram illustrates the differential induction of these various
tization to the drugs.
proteins by acute stimulation (1–2 hr after a single drug exposure) versus chronic In addition, there is preliminary evidence that the effects of
stimulation (1 day after repeated drug exposure). (B) Several waves of Fos-like ⌬FosB may extend well beyond a regulation of drug sensitivity
proteins [comprised of c-Fos (52- to 58-kDa isoforms), FosB (46- to 50-kDa iso- per se to more complex behaviors related to the addiction
forms), ⌬FosB (33-kDa isoform), and Fra1 or Fra2 (40 kDa)] are induced in nucleus process. Mice expressing ⌬FosB work harder to self-administer
accumbens and dorsal striatal neurons by acute administration of a drug of abuse. cocaine in progressive ratio self-administration assays, suggest-
Also induced are biochemically modified isoforms of ⌬FosB (35–37 kDa); they, ing that ⌬FosB may sensitize animals to the incentive motiva-
too, are induced (although at low levels) after acute drug administration, but
tional properties of cocaine and thereby lead to a propensity for
persist in brain for long periods because of their stability. (C) With repeated (e.g.,
twice daily) drug administration, each acute stimulus induces a low level of the
relapse after drug withdrawal.‡ ⌬FosB-expressing mice also
stable ⌬FosB isoforms, which is indicated by the lower set of overlapping lines that show enhanced anxiolytic effects of alcohol,§ a phenotype that
indicate ⌬FosB induced by each acute stimulus. The result is a gradual increase in has been associated with increased alcohol intake in humans.
the total levels of ⌬FosB with repeated stimuli during a course of chronic treat-
ment, which is indicated by the increasing stepped line in the graph.
‡Whisler, K., Kelz, M. B., Chen, J. S., Nestler, E. J. & Self, D. W. (1999) Soc. Neurosci. Abstr.
Biochemical Identity of Stable ⌬FosB Isoforms 25, 811.

As mentioned above, the ⌬FosB isoforms that accumulate after §Roberts, A., Picetti, R., Nestler, E. J., Koob, G. F. (2001) Soc. Neurosci. Abstr., in press.
chronic administration of a drug of abuse or compulsive running ¶Peakman, M.-C., Colby, C., Duman, R. S., Allen, M. R., Stock, J. L., NcNeish, J. D., Kelz, M. B.,

show a molecular mass of 35–37 kDa. They can be differentiated Chen, J. S., Nestler, E. J. & Schaeffer, E. (2000) Soc. Neurosci. Abstr. 26, 124.

Nestler et al. PNAS 兩 September 25, 2001 兩 vol. 98 兩 no. 20 兩 11043


Together, these early findings suggest that ⌬FosB, in addition to
increasing sensitivity to drugs of abuse, produces qualitative
changes in behavior that promote drug-seeking behavior. Thus,
⌬FosB may function as a sustained ‘‘molecular switch’’ that helps
initiate and then maintain crucial aspects of the addicted state.
An important question under current investigation is whether
⌬FosB accumulation during drug exposure promotes drug-
seeking behavior after extended withdrawal periods, even after
⌬FosB levels have normalized (see below).
Adult mice that overexpress ⌬FosB selectively within the
nucleus accumbens and dorsal striatum also exhibit greater
compulsive running compared with control littermates.† These
observations raise the interesting possibility that ⌬FosB accu-
mulation within these neurons serves a more general role in the
formation and maintenance of habit memories and compulsive
behaviors, perhaps by reinforcing the efficacy of neural circuits
in which those neurons function.
⌬FosB accumulates in certain brain regions outside the nu-
cleus accumbens and dorsal striatum after chronic exposure to
cocaine. Prominent among these regions are the amygdala and
medial prefrontal cortex (15). A major goal of current research Fig. 2. The AMPA glutamate receptor subunit, GluR2, is a putative target for
is to understand the contributions of ⌬FosB induction in these ⌬FosB. Shown is how ⌬FosB-mediated induction of GluR2 may alter the
regions to the addiction phenotype. physiological responsiveness of nucleus accumbens neurons and lead to sen-
sitized responses to drugs of abuse. According to this scheme, drugs of abuse
Earlier work on fosB knockout mice revealed that these
produce their acute reinforcing effects via inhibition of nucleus accumbens
animals fail to develop sensitization to the locomotor effects of neurons. With repeated exposure, the drugs induce ⌬FosB, which regulates
cocaine, which is consistent with the findings of the ⌬FosB- numerous target genes, including GluR2. This increases the proportion of
overexpressing mice mentioned above (22). However, the fosB AMPA receptors (AMPA-R) on nucleus accumbens neurons that contain the
mutants showed enhanced sensitivity to cocaine’s acute effects, GluR2 subunit, which causes reduced overall AMPA current and reduced Ca2⫹
which is inconsistent with these other findings. Interpretation of current. This reduced excitability could render the neurons more sensitive to
findings with the fosB mutants, though, is complicated by the fact the acute inhibitory effects of the drugs and thereby to the drugs’ reinforcing
that these animals lack not only ⌬FosB, but full-length FosB as effects.
well. Moreover, the mutants lack both proteins throughout the
brain and from the earliest stages of development. Indeed, more mutant attenuates the ability of cocaine to induce the protein.¶
recent work supports conclusions from the ⌬FosB overexpress- In addition, the promoter of the GluR2 gene contains a con-
ing mice: inducible overexpression of a truncated mutant of sensus AP-1 site that binds ⌬FosB (28). Overexpression of
c-Jun, which acts as a dominant negative antagonist of ⌬FosB, GluR2 in the nucleus accumbens, by use of viral-mediated gene
selectively in nucleus accumbens and dorsal striatum shows transfer, increases an animal’s sensitivity to the rewarding effects
reduced sensitivity to the rewarding effects of cocaine.¶ These of cocaine, thereby mimicking part of the phenotype seen in the
findings emphasize the caution that must be used in interpreting ⌬FosB-expressing mice (28). Induction of GluR2 could account
results from mice with constitutive mutations and illustrate the for the reduced electrophysiological sensitivity of nucleus ac-
importance of mice with inducible and cell type-specific muta- cumbens neurons to AMPA receptor agonists after chronic
tions in studies of plasticity in the adult brain. cocaine administration (32), because AMPA receptors contain-
ing GluR2 show reduced overall conductance and reduced Ca2⫹
Target Genes for ⌬FosB permeability. Reduced responsiveness of these neurons to exci-
Because ⌬FosB is a transcription factor, presumably the protein tatory inputs may then enhance responses to a drug of abuse.
causes behavioral plasticity through alterations in the expression However, the ways in which dopaminergic and glutamatergic
of other genes. ⌬FosB is generated by alternative splicing of the signals in nucleus accumbens regulate addictive behavior remain
fosB gene and lacks a portion of the C-terminal transactivation unknown; this will require a neural circuit level of understand-
domain present in full-length FosB. As a result, it was originally ing, which is not yet available.
proposed that ⌬FosB functions as a transcriptional repressor Another putative target for ⌬FosB is the gene encoding
(29). However, work in cell culture has demonstrated clearly that dynorphin. As stated earlier, dynorphin is expressed in the subset
⌬FosB can either induce or repress AP-1-mediated transcription of nucleus accumbens medium spiny neurons that show induc-
depending on the particular AP-1 site used (21, 29–31). Full- tion of ⌬FosB. Dynorphin appears to function in an intercellular
length FosB exerts the same effects as ⌬FosB on certain feedback loop: its release inhibits the dopaminergic neurons that
promoter fragments, but different effects on others. Further innervate the medium spiny neurons, via ␬ opioid receptors
work is needed to understand the mechanisms underlying these present on dopaminergic nerve terminals in the nucleus accum-
varied actions of ⌬FosB and FosB. bens and also on cell bodies and dendrites in the ventral
Our group has used two approaches to identify target genes for tegmental area (Fig. 3) (33–35). This idea is consistent with the
⌬FosB. One is the candidate gene approach. We initially con- ability of a ␬ receptor agonist, upon administration into either of
sidered ␣-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid these two brain regions, to decrease drug reward (35). Recent
(AMPA) glutamate receptors as putative targets, given the work has indicated that ⌬FosB decreases the expression of
important role of glutamatergic transmission in the nucleus dynorphin,储 which could contribute to the enhancement of
accumbens. Work to date has indicated that one particular reward mechanisms seen with ⌬FosB induction. Interestingly,
AMPA glutamate receptor subunit, GluR2, may be a bona fide another drug-regulated transcription factor, CREB (cAMP re-
target for ⌬FosB (Fig. 2). GluR2 expression, but not the sponse element binding protein) (2, 3), exerts the opposite
expression of other AMPA receptor subunits, is increased in
nucleus accumbens (but not dorsal striatum) upon overexpres-
sion of ⌬FosB (28), and expression of a dominant negative 储Shaw, T. Z., Gilden, L., Kelz, M., Chen, J. & Nestler, E. J. (2000) Soc. Neurosci. Abstr. 26, 525.

11044 兩 www.pnas.org兾cgi兾doi兾10.1073兾pnas.191352698 Nestler et al.


Fig. 3. Dynorphin is a putative target for ⌬FosB. Shown is a ventral tegmen- Fig. 4. Regulation of dendritic structure by drugs of abuse. Shown is the
tal area (VTA) dopamine (DA) neuron innervating a class of nucleus accum- expansion of a neuron’s dendritic tree after chronic exposure to a drug of
bens (NAc) GABAergic projection neuron that expresses dynorphin (DYN). abuse, as has been observed with cocaine in the nucleus accumbens and
Dynorphin serves a feedback mechanism in this circuit: dynorphin, released prefrontal cortex (41). The areas of magnification show an increase in den-
from terminals of the NAc neurons, acts on ␬ opioid receptors located on nerve dritic spines, which is postulated to occur in conjunction with activated nerve
terminals and cell bodies of the DA neurons to inhibit their functioning. ⌬FosB, terminals. This increase in dendritic spine density may be mediated via ⌬FosB
by inhibiting dynorphin expression, may down-regulate this feedback loop and the consequent induction of Cdk5 (see text). Such alterations in dendritic
and enhance the rewarding properties of drugs of abuse. Not shown is the structure, which are similar to those observed in some learning models (e.g.,
reciprocal effect of CREB on this system: CREB enhances dynorphin expression long-term potentiation), could mediate long-lived sensitized responses to
and thereby attenuates the rewarding properties of drugs of abuse (4). GABA, drugs of abuse or environmental cues. [Reproduced with permission from ref.
␥-aminobutyric acid; DR, dopamine receptor; OR, opioid receptor. 3 (Copyright 2001, Macmillian Magazines Ltd.)].

effect: it induces dynorphin expression in the nucleus accumbens The difficulty in finding the molecular adaptations that underlie
and reduces the rewarding properties of cocaine and morphine the extremely stable behavioral changes associated with addic-
(4).** Because drug-induced activation of CREB dissipates tion is analogous to the challenges faced in the learning and
rapidly after drug administration, such reciprocal regulation of memory field. Although there are elegant cellular and molecular
dynorphin by CREB and ⌬FosB could explain the reciprocal models of learning and memory, it has not to date been possible
behavioral changes that occur during early and late phases of to identify molecular and cellular adaptations that are suffi-
withdrawal, with negative emotional symptoms and reduced ciently long-lived to account for highly stable behavioral mem-
drug sensitivity predominating during early phases of with- ories. Indeed, ⌬FosB is the longest-lived adaptation known to
drawal, and sensitization to the rewarding and incentive moti- occur in adult brain, not only in response to drugs of abuse, but
vational effects of drugs predominating at later time points. to any other perturbation (that doesn’t involve lesions) as well.
The second approach used to identify target genes for ⌬FosB Two proposals have evolved, both in the addiction and learning
involves DNA microarray analysis. Inducible overexpression of and memory fields, to account for this discrepancy.
⌬FosB increases or decreases the expression of numerous genes

COLLOQUIUM
One possibility is that more transient changes in gene
in the nucleus accumbens (36). Although considerable work is expression, such as those mediated via ⌬FosB or other tran-
now needed to validate each of these genes as physiologic targets
scription factors (e.g., CREB), may mediate more long-lived
of ⌬FosB and to understand their contribution to the addiction
changes in neuronal morphology and synaptic structure. For
phenotype, one important target appears to be Cdk5 (cyclin-
example, an increase in the density of dendritic spines (par-
dependent kinase-5). Thus, Cdk5 was initially identified as
ticularly an increase in two-headed spines) accompanies the
⌬FosB-regulated by use of microarrays, and later shown to be
increased efficacy of glutamatergic synapses at hippocampal
induced in nucleus accumbens and dorsal striatum after chronic
cocaine administration (37). ⌬FosB activates the cdk5 gene via pyramidal neurons during long-term potentiation (38 – 40),
an AP-1 site present within the gene’s promoter (36). Together, and parallels the enhanced behavioral sensitivity to cocaine
these data support a scheme wherein cocaine induces Cdk5 mediated at the level of medium spiny neurons of the nucleus
expression in these brain regions via ⌬FosB. Induction of Cdk5 accumbens (41). It is not known whether such structural
appears to alter dopaminergic signaling at least in part via changes are sufficiently long-lived to account for highly stable
increased phosphorylation of DARPP-32 (37), which is con- changes in behavior, although the latter persist for at least 1
verted from an inhibitor of protein phosphatase-1 to an inhibitor month of drug withdrawal. Recent evidence raises the possi-
of protein kinase A upon its phosphorylation by Cdk5 (26). bility that ⌬FosB, and its induction of Cdk5, is one mediator
of drug-induced changes in synaptic structure in the nucleus
Role of ⌬FosB in Mediating ‘‘Permanent’’ Plasticity to Drugs accumbens (Fig. 4).†† Thus, infusion of a Cdk5 inhibitor into
of Abuse the nucleus accumbens prevents the ability of repeated cocaine
Although the ⌬FosB signal is relatively long-lived, it is not exposure to increase dendritic spine density in this region. This
permanent. ⌬FosB degrades gradually and can no longer be is consistent with the view that Cdk5, which is enriched in
detected in brain after 1–2 months of drug withdrawal, even brain, regulates neural structure and growth (see refs. 36 and
though certain behavioral abnormalities persist for much longer 37). It is possible, although by no means proven, that such
periods of time. Therefore, ⌬FosB per se would not appear to be changes in neuronal morphology may outlast the ⌬FosB signal
able to mediate these semipermanent behavioral abnormalities. itself.

**Barrot, M., Olivier, J. D. A., Zachariou, V., Neve, R. L. & Nestler, E. J. (2000) Soc. Neurosci. ††Norrholm, S.D., Bibb, J. A., Nestler, E. J., Ouimet, C. C., Taylor, J. R. & Greengard, P. (2001)
Abstr. 26, 485. Soc. Neurosci. Abstr., in press.

Nestler et al. PNAS 兩 September 25, 2001 兩 vol. 98 兩 no. 20 兩 11045


Another possibility is that the transient induction of a tran- longer-lived changes in gene expression by regulating other
scription factor (e.g., ⌬FosB, CREB) leads to more permanent modifications of chromatin (e.g., DNA or histone methylation)
changes in gene expression through the modification of chro- that have been implicated in the permanent changes in gene
matin. These and many other transcription factors are believed transcription that occur during development (see refs. 42 and
to activate or repress the transcription of a target gene by 43). Although these possibilities remain speculative, they could
promoting the acetylation or deacetylation, respectively, of provide a mechanism by which transient adaptations to a drug
histones in the vicinity of the gene (42). Although such acety- of abuse (or some other perturbation) lead to essentially life-long
lation and deacetylation of histones can apparently occur very behavioral consequences.
rapidly, it is possible that ⌬FosB or CREB might produce
longer-lasting adaptations in the enzymatic machinery that This work was supported by grants from the National Institute on Drug
controls histone acetylation. ⌬FosB or CREB may also promote Abuse.

1. Nestler, E. J., Hope, B. T. & Widnell, K. L. (1993) Neuron 11, 995–1006. 24. Hiroi, N., Feinberg, A., Haile, C., Greengard, P. & Nestler, E. J. (1999) Eur.
2. Berke, J. D. & Hyman, S. E. (2000) Neuron 25, 515–532. J. Neurosci. 11, 1114–1118.
3. Nestler, E. J. (2001) Nat. Rev. Neurosci. 2, 119–128. 25. Greengard, P., Allen, P. B. & Nairn, A. C. (1999) Neuron 23, 435–447.
4. Carlezon, W. A., Jr., Thome, J., Olson, V. G., Lane-Ladd, S. B., Brodkin, E. S., 26. Bibb, J. A., Snyder, G. L., Nishi, A., Yan, Z., Meijer, L., Fienberg, A. A., Tsai,
Hiroi, N., Duman, R. S., Neve, R. L. & Nestler, E. J. (1998) Science 282, L. H., Kwon, Y. T., Girault, J. A., Czernik, A. J., et al. (1999) Nature (London)
2272–2275. 402, 669–671.
5. O’Donovan, K. J., Tourtellotte, W. G., Millbrandt, J. & Baraban, J. M. (1999) 27. Chen, J. S., Kelz, M. B., Zeng, G. Q., Sakai, N., Steffen, C., Shockett, P. E.,
Trends Neurosci. 22, 167–173. Picciotto, M., Duman, R. S. & Nestler, E. J. (1998) Mol. Pharmacol. 54,
6. Mackler, S. A., Korutla, L., Cha, X. Y., Koebbe, M. J., Fournier, K. M., Bowers,
495–503.
M. S. & Kalivas, P. W. (2000) J. Neurosci. 20, 6210–6217.
28. Kelz, M. B., Chen, J. S., Carlezon, W. A., Whisler, K., Gilden, L., Beckmann,
7. Morgan, J. I. & Curran, T. (1995) Trends Neurosci. 18, 66–67.
A. M., Steffen, C., Zhang, Y.-J., Marotti, L., Self, S. W., et al. (1999) Nature
8. Young, S. T., Porrino, L. J. & Iadarola, M. J. (1991) Proc. Natl. Acad. Sci. USA
88, 1291–1295. (London) 401, 272–276.
9. Graybiel, A. M., Moratalla, R. & Robertson, H. A. (1990) Proc. Natl. Acad. Sci. 29. Dobrazanski, P., Noguchi, T., Kovary, K., Rizzo, C. A., Lazo, P. S. & Bravo,
USA 87, 6912–6916. R. (1991) Mol. Cell Biol. 11, 5470–5478.
10. Hope, B., Kosofsky, B., Hyman, S. E. & Nestler, E. J. (1992) Proc. Natl. Acad. 30. Nakabeppu, Y. & Nathans, D. (1991) Cell 64, 751–759.
Sci. USA 89, 5764–5768. 31. Yen, J., Wisdom, R. M., Tratner, I. & Verma, I. M. (1991) Proc. Natl. Acad.
11. Kelz, M. B. & Nestler, E. J. (2000) Curr. Opin. Neurol. 13, 715–720. Sci. USA 88, 5077–5081.
12. Koob, G. F., Sanna, P. P. & Bloom, F. E. (1998) Neuron 21, 467–476. 32. White, F. J., Hu, X.-T., Zhang, X.-F. & Wolf, M. E. (1995) J. Pharmacol. Exp.
13. Wise, R. A. (1998) Drug Alcohol Dependence 51, 13–22. Ther. 273, 445–454.
14. Hope, B. T., Nye, H. E., Kelz, M. B., Self, D. W., Iadarola, M. J., Nakabeppu, 33. Hyman, S. E. (1996) Neuron 16, 901–904.
Y., Duman, R. S. & Nestler, E. J. (1994) Neuron 13, 1235–1244. 34. Kreek, M. J. (1997) Pharmacol. Biochem. Behav. 57, 551–569.
15. Nye, H., Hope, B. T., Kelz, M., Iadarola, M. & Nestler, E. J. (1995) 35. Shippenberg, T. S. & Rea, W. (1997) Pharmacol. Biochem. Behav. 57,
J. Pharmacol. Exp. Ther. 275, 1671–1680. 449 – 455.
16. Nye, H. E. & Nestler, E. J. (1996) Mol. Pharmacol. 49, 636–645. 36. Chen, J. S., Zhang, Y. J., Kelz, M. B., Steffen, C., Ang, E. S., Zeng, L. & Nestler,
17. Moratalla, R., Elibol, B., Vallejo, M. & Graybiel, A. M. (1996) Neuron 17, 147–156. E. J. (2000) J. Neurosci. 20, 8965–8971.
18. Pich, E. M., Pagliusi, S. R., Tessari, M., Talabot-Ayer, D., Hooft van Huijs- 37. Bibb, J. A., Chen, J. S., Taylor, J. R., Svenningsson, P., Nishi, A., Snyder, G. L.,
duijnen, R. & Chiamulera, C. (1997) Science 275, 83–86. Yan, Z., Sagawa, Z. K., Nairn, A. C., Nestler, E. J., et al. (2001) Nature (London)
19. Chen, J. S., Nye, H. E., Kelz, M. B., Hiroi, N., Nakabeppu, Y., Hope, B. T. &
410, 376–380.
Nestler, E. J. (1995) Mol. Pharmacol. 48, 880–889.
38. Luscher, C., Nicoll, R. A., Malenka, R. C. & Muller, D. (2000) Nat. Neurosci.
20. Hiroi, N., Brown, J., Ye, H., Saudou, F., Vaidya, V. A., Duman, R. S.,
3, 545–550.
Greenberg, M. E. & Nestler, E. J. (1998) J. Neurosci. 18, 6952–6962.
21. Chen, J., Kelz, M. B., Hope, B. T., Nakabeppu, Y. & Nestler, E. J. (1997) 39. Malinow, R., Mainen, Z. F. & Hayashi, Y. (2000) Curr. Opin. Neurobiol. 10,
J. Neurosci. 17, 4933–4941. 352–357.
22. Hiroi, N., Brown, J., Haile, C., Ye, H., Greenberg, M. E. & Nestler, E. J. (1997) 40. Scannevin, R. H. & Huganir, R. L. (2000) Nat. Rev. Neurosci. 1, 133–141.
Proc. Natl. Acad. Sci. USA 94, 10397–10402. 41. Robinson, T. E. & Kolb, B. (1999) (1997) Eur. J. Neurosci. 11, 1598–1604.
23. Fienberg, A. A., Hiroi, N., Mermelstein, P., Song, W.-J., Snyder, G. L., Nishi, 42. Carey, M. & Smale, S. T. (2000) Transcriptional Regulation in Eukaryotes (Cold
A., Cheramy, A., O’Callaghan, J. P., Miller, D., Cole, D. G., et al. (1998) Science Spring Harbor Lab. Press, Plainview, NY).
281, 838–842. 43. Spencer, V. A. & Davie, J. R. (1999) Gene 240, 1–12.

11046 兩 www.pnas.org兾cgi兾doi兾10.1073兾pnas.191352698 Nestler et al.

You might also like